1
|
Bonfanti L, Couillard-Després S. Neuron and Brain Maturation 2.0. Int J Mol Sci 2023; 24:17113. [PMID: 38069434 PMCID: PMC10707153 DOI: 10.3390/ijms242317113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The mammalian central nervous system (CNS) is built up during embryogenesis by neural stem cells located in the periventricular germinal layers which undergo multiple division cycles [...].
Collapse
Affiliation(s)
- Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), 10043 Orbassano, Italy
- Department of Veterinary Sciences, University of Turin, 10095 Torino, Italy
| | - Sébastien Couillard-Després
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
2
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
3
|
Abdullatef S, Farina C. Publicly available ex vivo transcriptomics datasets to explore CNS physiology and neurodegeneration: state of the art and perspectives. Front Neurosci 2023; 17:1211079. [PMID: 37680966 PMCID: PMC10481165 DOI: 10.3389/fnins.2023.1211079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
The central nervous system (CNS) is characterized by an intricate composition of diverse cell types, including neurons and glia cells (astrocytes, oligodendrocytes, and microglia), whose functions may differ along time, between sexes and upon pathology. The advancements in high-throughput transcriptomics are providing fundamental insights on cell phenotypes, so that molecular codes and instructions are ever more described for CNS physiology and neurodegeneration. To facilitate the search of relevant information, this review provides an overview of key CNS transcriptomics studies ranging from CNS development to ageing and from physiology to pathology as defined for five neurodegenerative disorders and their relative animal models, with a focus on molecular descriptions whose raw data were publicly available. Accurate phenotypic descriptions of cellular states correlate with functional changes and this knowledge may support research devoted to the development of therapeutic strategies supporting CNS repair and function.
Collapse
Affiliation(s)
- Sandra Abdullatef
- Division of Neuroscience, Institute of Experimental Neurology (INSpe), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Cinthia Farina
- Division of Neuroscience, Institute of Experimental Neurology (INSpe), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
4
|
Gao Y, Syed M, Zhao X. Mechanisms underlying the effect of voluntary running on adult hippocampal neurogenesis. Hippocampus 2023; 33:373-390. [PMID: 36892196 PMCID: PMC10566571 DOI: 10.1002/hipo.23520] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
Adult hippocampal neurogenesis is important for preserving learning and memory-related cognitive functions. Physical exercise, especially voluntary running, is one of the strongest stimuli to promote neurogenesis and has beneficial effects on cognitive functions. Voluntary running promotes exit of neural stem cells (NSCs) from the quiescent stage, proliferation of NSCs and progenitors, survival of newborn cells, morphological development of immature neuron, and integration of new neurons into the hippocampal circuitry. However, the detailed mechanisms driving these changes remain unclear. In this review, we will summarize current knowledge with respect to molecular mechanisms underlying voluntary running-induced neurogenesis, highlighting recent genome-wide gene expression analyses. In addition, we will discuss new approaches and future directions for dissecting the complex cellular mechanisms driving change in adult-born new neurons in response to physical exercise.
Collapse
Affiliation(s)
- Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Moosa Syed
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
5
|
Hourigan B, Balay SD, Yee G, Sharma S, Tan Q. Capicua regulates the development of adult-born neurons in the hippocampus. Sci Rep 2021; 11:11725. [PMID: 34083623 PMCID: PMC8175746 DOI: 10.1038/s41598-021-91168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 05/20/2021] [Indexed: 11/12/2022] Open
Abstract
New neurons continuously arise from neural progenitor cells in the dentate gyrus of the adult hippocampus to support ongoing learning and memory formation. To generate functional adult-born neurons, neural progenitor cells proliferate to expand the precursor cell pool and differentiate into neurons. Newly generated cells then undergo postmitotic maturation to migrate to their final destination and develop elaborate dendritic branching, which allows them to receive input signals. Little is known about factors that regulate neuronal differentiation, migration, and dendrite maturation during adult hippocampal neurogenesis. Here, we show that the transcriptional repressor protein capicua (CIC) exhibits dynamic expression in the adult dentate gyrus. Conditional deletion of Cic from the mouse dentate gyrus compromises the adult neural progenitor cell pool without altering their proliferative potential. We further demonstrate that the loss of Cic impedes neuronal lineage development and disrupts dendritic arborization and migration of adult-born neurons. Our study uncovers a previously unrecognized role of CIC in neurogenesis of the adult dentate gyrus.
Collapse
Affiliation(s)
- Brenna Hourigan
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Spencer D Balay
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada.,Research Institute of Molecular Pathology, Vienna Biocenter, Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Graydon Yee
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Saloni Sharma
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, T6J 2H7, Canada.
| |
Collapse
|
6
|
Gao Y, Shen M, Gonzalez JC, Dong Q, Kannan S, Hoang JT, Eisinger BE, Pandey J, Javadi S, Chang Q, Wang D, Overstreet-Wadiche L, Zhao X. RGS6 Mediates Effects of Voluntary Running on Adult Hippocampal Neurogenesis. Cell Rep 2021; 32:107997. [PMID: 32755589 DOI: 10.1016/j.celrep.2020.107997] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 01/17/2023] Open
Abstract
Voluntary running enhances adult hippocampal neurogenesis, with consequences for hippocampal-dependent learning ability and mood regulation. However, the underlying mechanism remains unclear. Here, we show that voluntary running induces unique and dynamic gene expression changes specifically within the adult-born hippocampal neurons, with significant impact on genes involved in neuronal maturation and human diseases. We identify the regulator of G protein signaling 6 (RGS6) as a key factor that mediates running impact on adult-born neurons. RGS6 overexpression mimics the positive effects of voluntary running on morphological and physiological maturation of adult new neurons and reduced sensitivity of adult-born neurons to the inhibitory effect of GABAB (γ-Aminobutyric acid B) receptor activation. Knocking down RGS6 abolishes running-enhanced neuronal maturation and hippocampal neurogenesis-dependent learning and anxiolytic effect. Our study provides a data resource showing genome-wide intrinsic molecular changes in adult-born hippocampal neurons that contribute to voluntary running-induced neurogenesis.
Collapse
Affiliation(s)
- Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sudharsan Kannan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Johnson T Hoang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian E Eisinger
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jyotsna Pandey
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sahar Javadi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
7
|
Wang X, Ding R, Song Y, Wang J, Zhang C, Han S, Han J, Zhang R. Transcutaneous Electrical Acupoint Stimulation in Early Life Changes Synaptic Plasticity and Improves Symptoms in a Valproic Acid-Induced Rat Model of Autism. Neural Plast 2020; 2020:8832694. [PMID: 33456456 PMCID: PMC7787794 DOI: 10.1155/2020/8832694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/12/2020] [Indexed: 01/30/2023] Open
Abstract
Autism spectrum disorder (ASD) is a developmental disorder characterized by social behavior deficit in childhood without satisfactory medical intervention. Transcutaneous electrical acupoint stimulation (TEAS) is a noninvasive technique derived from acupuncture and has been shown to have similar therapeutic effects in many diseases. Valproic acid- (VPA-) induced ASD is a known model of ASD in rats. The therapeutic efficacy of TEAS was evaluated in the VPA model of ASD in the present study. The offspring of a VPA-treated rat received TEAS in the early life stage followed by a series of examinations conducted in their adolescence. The results show that following TEAS treatment in early life, the social and cognitive ability in adolescence of the offspring of a VPA rat were significantly improved. In addition, the abnormal pain threshold was significantly corrected. Additional studies demonstrated that the dendritic spine density of the primary sensory cortex was decreased with Golgi staining. Results of the transcriptomic study showed that expression of some transcription factors such as the neurotrophic factor were downregulated in the hypothalamus of the VPA model of ASD. The reduced gene expression was reversed following TEAS. These results suggest that TEAS in the early life stage may mitigate disorders of social and recognition ability and normalize the pain threshold of the ASD rat model. The mechanism involved may be related to improvement of synaptic plasticity.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Rui Ding
- Department of Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yayue Song
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Juan Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Songping Han
- Wuxi HANS Health Medical Technology Co., Ltd., Wuxi, China
| | - Jisheng Han
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
8
|
Abstract
The mammalian brain has over 10,000 types of neurons. Therefore, studying gene regulation in the brain requires effective strategies for targeting specific cell types, especially those in low abundance. Cell isolation may alter gene expression and is disruptive to mature neurons with extensive processes. This protocol describes cell-type-specific expression of tagged ribosome and the use of ribosome tagging followed by RNA-seq to identify translatome of low number and sparse cells in mouse brains without disruptive cell isolation. For complete details on the use and execution of this protocol, please refer to Gao et al. (2020).
Collapse
|
9
|
Park DI. Genomics, transcriptomics, proteomics and big data analysis in the discovery of new diagnostic markers and targets for therapy development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:61-90. [PMID: 32711818 DOI: 10.1016/bs.pmbts.2020.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Highly complex endophenotypes and underlying molecular mechanisms have prevented effective diagnosis and treatment of autism spectrum disorder. Despite extensive studies to identify relevant biosignatures, no biomarker and therapeutic targets are available in the current clinical practice. While our current knowledge is still largely incomplete, -omics technology and machine learning-based big data analysis have provided novel insights on the etiology of autism spectrum disorders, elucidating systemic impairments that can be translated into biomarker and therapy target candidates. However, more integrated and sophisticated approaches are vital to realize molecular stratification and individualized treatment strategy. Ultimately, systemic approaches based on -omics and big data analysis will significantly contribute to more effective biomarker and therapy development for autism spectrum disorder.
Collapse
Affiliation(s)
- Dong Ik Park
- Danish Research Institute of Translational Neuroscience (DANDRITE)-Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Danish National Research Foundation Center, PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Vitriolo A, Gabriele M, Testa G. From enhanceropathies to the epigenetic manifold underlying human cognition. Hum Mol Genet 2019; 28:R226-R234. [PMID: 31411680 PMCID: PMC6990140 DOI: 10.1093/hmg/ddz196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/27/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
A vast portion of intellectual disability and autism spectrum disorders is genetically caused by mutations in chromatin modulators. These proteins play key roles in development and are also highly expressed in the adult brain. Specifically, the pivotal role of chromatin regulation in transcription has placed enhancers at the core of neurodevelopmental disorders (NDDs) studies, ushering in the coining of the term enhanceropathies. The convergence of these disorders is multilayered, spanning from molecular causes to pathophysiological traits, including extensive overlaps between enhanceropathies and neurocristopathies. The reconstruction of epigenetic circuitries wiring development and underlying cognitive functions has gone hand in hand with the development of tools that increase the sensitivity of identifying regulatory regions and linking enhancers to their target genes. The available models, including loop extrusion and phase separation, have been bringing into relief complementary aspects to interpret gene regulation datasets, reinforcing the idea that enhancers are not all the same and that regulatory regions possess shades of enhancer-ness and promoter-ness. The current limits in enhancer definition, within the emerging broader understanding of chromatin dynamics in time and space, are now on the verge of being transformed by the possibility to interrogate developmentally relevant three-dimensional cellular models at single-cell resolution. Here we discuss the contours of how these technological advances, as well as the epistemic limitations they are set to overcome, may well usher in a change of paradigm for NDDs, moving the quest for convergence from enhancers to the four-dimensional (4D) genome.
Collapse
Affiliation(s)
- Alessandro Vitriolo
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
11
|
Mehmood S, Bilal M, Manzoor R, Iqbal H. Deciphering the adult brain development complexity by single-cell transcriptome analysis—a review. MATERIALS TODAY CHEMISTRY 2019. [DOI: 10.1016/j.mtchem.2019.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
12
|
Sun Y, Gao Y, Tidei JJ, Shen M, Hoang JT, Wagner DF, Zhao X. Loss of MeCP2 in immature neurons leads to impaired network integration. Hum Mol Genet 2019; 28:245-257. [PMID: 30277526 DOI: 10.1093/hmg/ddy338] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations or deletions in Methyl-CpG-binding Protein 2 (MeCP2), a brain-enriched transcriptional regulator. MeCP2 is highly expressed during neuronal maturation and its deficiency results in impaired dendritic morphogenesis and reduced dendritic spine numbers in developing neurons. However, whether MeCP2 deficiency impacts the integration of new neurons has not been directly assessed. In this study, we developed a modified rabies virus-mediated monosynaptic retrograde tracing method to interrogate presynaptic integration of MeCP2-deficient new neurons born in the adult hippocampus, a region with lifelong neurogenesis and plasticity. We found that selective deletion of MeCP2 in adult-born new neurons impaired their long-range connectivity to the cortex, whereas their connectivity within the local hippocampal circuits or with subcortical regions was not significantly affected. We further showed that knockdown of MeCP2 in primary hippocampal neurons also resulted in reduced network integration. Interestingly, (1-3) insulin-like growth factor-1 (IGF-1), a small peptide under clinical trial testing for RTT, rescued neuronal integration deficits of MeCP2-deficient neurons in vitro but not in vivo. In addition, (1-3) IGF-1 treatment corrected aberrant excitability and network synchrony of MeCP2-deficient hippocampal neurons. Our results indicate that MeCP2 is essential for immature neurons to establish appropriate network connectivity.
Collapse
Affiliation(s)
- Yi Sun
- National Key Research Laboratory of Natural and Biomimetic Drugs.,Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, PR China.,Waisman Center
| | - Yu Gao
- Waisman Center.,Department of Neuroscience
| | | | | | | | | | - Xinyu Zhao
- Waisman Center.,Department of Neuroscience.,Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
13
|
Reduced mitochondrial fusion and Huntingtin levels contribute to impaired dendritic maturation and behavioral deficits in Fmr1-mutant mice. Nat Neurosci 2019; 22:386-400. [PMID: 30742117 PMCID: PMC6556892 DOI: 10.1038/s41593-019-0338-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/09/2019] [Indexed: 12/26/2022]
Abstract
Fragile X syndrome results from a loss of the RNA-binding protein fragile X mental retardation protein (FMRP). How FMRP regulates neuronal development and function remains unclear. Here we show that FMRP-deficient immature neurons exhibit impaired dendritic maturation, altered expression of mitochondrial genes, fragmented mitochondria, impaired mitochondrial function, and increased oxidative stress. Enhancing mitochondrial fusion partially rescued dendritic abnormalities in FMRP-deficient immature neurons. We show that FMRP deficiency leads to reduced Htt mRNA and protein levels and that HTT mediates FMRP regulation of mitochondrial fusion and dendritic maturation. Mice with hippocampal Htt knockdown and Fmr1-knockout mice showed similar behavioral deficits that could be rescued by treatment with a mitochondrial fusion compound. Our data unveil mitochondrial dysfunction as a contributor to the impaired dendritic maturation of FMRP-deficient neurons and suggest a role for interactions between FMRP and HTT in the pathogenesis of fragile X syndrome.
Collapse
|
14
|
Liu JYW, Matarin M, Reeves C, McEvoy AW, Miserocchi A, Thompson P, Sisodiya SM, Thom M. Doublecortin-expressing cell types in temporal lobe epilepsy. Acta Neuropathol Commun 2018; 6:60. [PMID: 30005693 PMCID: PMC6045867 DOI: 10.1186/s40478-018-0566-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022] Open
Abstract
Doublecortin (DCX) is widely regarded as a marker of immature and migrating neurons during development. While DCX expression persists in adults, particularly in the temporal lobe and neurogenic regions, it is unknown how seizures influence its expression. The aim of the present study was to explore the distribution and characteristics of DCX-expressing cells in surgical and postmortem samples from 40 adult and paediatric patients, with epilepsy and with or without hippocampal sclerosis (HS), compared to post mortem controls. The hippocampus (pes and body), parahippocampal gyrus, amygdala, temporal pole and temporal cortex were examined with DCX immunohistochemistry using four commercially-available DCX antibodies, labelled cells were quantified in different regions of interest as well as their co-expression with cell type specific markers (CD68, Iba1, GFAP, GFAP∂, nestin, SOX2, CD34, OLIG2, PDGFRβ, NeuN) and cell cycle marker (MCM2). Histological findings were compared with clinical data, as well as gene expression data obtained from the temporal cortex of 83 temporal lobe epilepsy cases with HS. DCX immunohistochemistry identified immature (Nestin−/NeuN−) neurons in layer II of the temporal neocortex in patients with and without epilepsy. Their number declined significantly with age but was not associated with the presence of hippocampal sclerosis, seizure semiology or memory dysfunction. DCX+ cells were prominent in the paralaminar nuclei and periamygdalar cortex and these declined with age but were not significantly associated with epilepsy history. DCX expressing cells with ramified processes were prominent in all regions, particularly in the hippocampal subgranular zone, where significantly increased numbers were observed in epilepsy samples compared to controls. DCX ramified cells co-expressed Iba1, CD68 and PDGFRβ, and less frequently MCM2, OLIG2 and SOX2, but no co-localization was observed with CD34, nestin or GFAP/GFAP ∂. Gene expression data from neocortical samples in patients with TLE and HS supported ongoing DCX expression in adults. We conclude that DCX identifies a range of morphological cell types in temporal lobe epilepsy, including immature populations, glial and microglial cell types. Their clinical relevance and biological function requires further study but we show some evidence for alteration with age and in epilepsy.
Collapse
|
15
|
Asymmetric Golgi Repositioning: A Prerequisite for Appropriate Dendrite Formation in Adult-Born Neurons. J Neurosci 2018; 38:4843-4845. [PMID: 29793931 DOI: 10.1523/jneurosci.0416-18.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/13/2018] [Accepted: 04/24/2018] [Indexed: 11/21/2022] Open
|
16
|
Harris L, Zalucki O, Clément O, Fraser J, Matuzelski E, Oishi S, Harvey TJ, Burne THJ, Heng JIT, Gronostajski RM, Piper M. Neurogenic differentiation by hippocampal neural stem and progenitor cells is biased by NFIX expression. Development 2018; 145:145/3/dev155689. [DOI: 10.1242/dev.155689] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 12/22/2017] [Indexed: 12/29/2022]
Abstract
ABSTRACT
Our understanding of the transcriptional programme underpinning adult hippocampal neurogenesis is incomplete. In mice, under basal conditions, adult hippocampal neural stem cells (AH-NSCs) generate neurons and astrocytes, but not oligodendrocytes. The factors limiting oligodendrocyte production, however, remain unclear. Here, we reveal that the transcription factor NFIX plays a key role in this process. NFIX is expressed by AH-NSCs, and its expression is sharply upregulated in adult hippocampal neuroblasts. Conditional ablation of Nfix from AH-NSCs, coupled with lineage tracing, transcriptomic sequencing and behavioural studies collectively reveal that NFIX is cell-autonomously required for neuroblast maturation and survival. Moreover, a small number of AH-NSCs also develop into oligodendrocytes following Nfix deletion. Remarkably, when Nfix is deleted specifically from intermediate progenitor cells and neuroblasts using a Dcx-creERT2 driver, these cells also display elevated signatures of oligodendrocyte gene expression. Together, these results demonstrate the central role played by NFIX in neuroblasts within the adult hippocampal stem cell neurogenic niche in promoting the maturation and survival of these cells, while concomitantly repressing oligodendrocyte gene expression signatures.
Collapse
Affiliation(s)
- Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Olivier Clément
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia 6102
| | - James Fraser
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Elise Matuzelski
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Sabrina Oishi
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Tracey J. Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia 4072
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Queensland, Australia 4076
| | - Julian Ik-Tsen Heng
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia 6102
| | - Richard M. Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia 4072
| |
Collapse
|
17
|
Eisinger BE, Zhao X. Identifying molecular mediators of environmentally enhanced neurogenesis. Cell Tissue Res 2018; 371:7-21. [PMID: 29127518 PMCID: PMC5826587 DOI: 10.1007/s00441-017-2718-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/16/2017] [Indexed: 02/06/2023]
Abstract
Adult hippocampal neurogenesis occurs throughout life and supports healthy brain functions. The production of new neurons decreases with age, and deficiencies in adult neurogenesis are associated with neurodevelopmental and degenerative disease. The rate of neurogenesis is dynamically sensitive to an individual's environmental conditions and experiences, and certain stimuli are known robustly to enhance neurogenesis in rodent models, including voluntary exercise, enriched environment, and electroconvulsive shock. In these models, information about an organism's environment and physiological state are relayed to neurogenic cell types within the hippocampus through a series of tissue and cellular interfaces, ultimately eliciting a neurogenic response from neural stem cells and newborn neurons. Therefore, an understanding of the way that novel genes and proteins act in specific cell types within this circuit-level context is of scientific and therapeutic value. Several well-studied neurotrophic factors have been implicated in environmentally enhanced neurogenesis. This review highlights recently discovered, novel molecular mediators of neurogenesis in response to environmental cues and summarizes the contribution of advanced, large-scale gene expression and function assessment technology to past, present, and future efforts aimed at elucidating cell-type-specific molecular mediators of environmentally enhanced neurogenesis.
Collapse
Affiliation(s)
- Brian E Eisinger
- Waisman Center and Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Xinyu Zhao
- Waisman Center and Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
18
|
Liu J, Reeves C, Jacques T, McEvoy A, Miserocchi A, Thompson P, Sisodiya S, Thom M. Nestin-expressing cell types in the temporal lobe and hippocampus: Morphology, differentiation, and proliferative capacity. Glia 2018; 66:62-77. [PMID: 28925561 PMCID: PMC5724502 DOI: 10.1002/glia.23211] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/30/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022]
Abstract
Nestin is expressed in immature neuroepithelial and progenitor cell types and transiently upregulated in proliferative neuroglial cells responding to acute brain injury, including following seizures. In 36 temporal lobe (TLobe) specimens from patients with TLobe epilepsy (age range 8-60 years) we studied the number, distribution and morphology of nestin-expressing cells (NEC) in the pes, hippocampus body, parahippocampal gyrus, amygdala, temporal cortex and pole compared with post mortem control tissues from 26 cases (age range 12 gestational weeks to 76 years). The proliferative fraction of NEC was evaluated in selected regions, including recognized niches, using MCM2. Their differentiation was explored with neuronal (DCX, mushashi, βIII tubulin, NeuN) and glial (GFAP, GFAPdelta, glutamine synthetase, aquaporin4, EAAT1) markers, both in sections or following culture. Findings were correlated with clinical parameters. A stereotypical pattern in the distribution and morphologies of NEC was observed, reminiscent of patterns in the developing brain, with increased densities in epilepsy than adult controls (p < .001). Findings included MCM2-positive radial glial-like cells in the periventricular white matter and rows of NEC in the hippocampal fimbria and sulcus. Nestin cells represented 29% of the hippocampal proliferative fraction in epilepsy cases; 20% co-expressed βIII tubulin in culture compared with 28% with GFAP. Significant correlations were noted between age at surgery, memory deficits and nestin populations. TLobe NEC with ongoing proliferative capacity likely represent vestiges of developmental migratory streams and resident reactive cell populations of potential relevance to hippocampal epileptogenesis, TLobe pathology, and co-morbidities, including memory decline.
Collapse
Affiliation(s)
- Joan Liu
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
- Department of Biomedical SciencesUniversity of WestminsterLondonW1W 6UWUnited Kingdom
| | - Cheryl Reeves
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Thomas Jacques
- Department of NeuropathologyUCL Institute of Child Health and Great Ormond Street Hospital for ChildrenLondonUnited Kingdom
| | - Andrew McEvoy
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Anna Miserocchi
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Pamela Thompson
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeuropsychologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Sanjay Sisodiya
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeurologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Maria Thom
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| |
Collapse
|
19
|
Abstract
Currently, the study of the transcriptome is widely used to interpret the functional elements of the genome and molecular constituents of cells and tissues in an effort to unravel biological pathways associated with development and disease. The advent of technologies is now enabling the study of such comprehensive transcriptional characterization of mRNA, miRNA, lncRNA, and small RNA in a robust and successful manner. Transcriptomic strategies are gaining momentum across diverse areas of biological, plant sciences, medical, clinical, and pharmaceutical research for biomarker discovery, and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Nalini Raghavachari
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, Bethesda, MD, USA.
| | - Natàlia Garcia-Reyero
- Environmental Laboratory, US Army Engineer Research and Development Center, Vicksburg, MS, USA.
| |
Collapse
|
20
|
Repositioning of Somatic Golgi Apparatus Is Essential for the Dendritic Establishment of Adult-Born Hippocampal Neurons. J Neurosci 2017; 38:631-647. [PMID: 29217690 DOI: 10.1523/jneurosci.1217-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/02/2017] [Accepted: 10/29/2017] [Indexed: 01/22/2023] Open
Abstract
New dentate granule cells (DGCs) are continuously generated, and integrate into the preexisting hippocampal network in the adult brain. How an adult-born neuron with initially simple spindle-like morphology develops into a DGC, consisting of a single apical dendrite with further branches, remains largely unknown. Here, using retroviruses to birth date and manipulate newborn neurons, we examined initial dendritic formation and possible underlying mechanisms. We found that GFP-expressing newborn cells began to establish a DGC-like morphology at ∼7 d after birth, with a primary dendrite pointing to the molecular layer, but at this stage, with several neurites in the neurogenic zone. Interestingly, the Golgi apparatus, an essential organelle for neurite growth and maintenance, was dynamically repositioning in the soma of newborn cells during this initial integration stage. Two weeks after birth, by which time most neurites in the neurogenic zone were eliminated, a compact Golgi apparatus was positioned exclusively at the base of the primary dendrite. We analyzed the presence of Golgi-associated genes using single-cell transcriptomes of newborn DGCs, and among Golgi-related genes, found the presence of STK25 and STRAD, regulators of embryonic neuronal development. When we knocked down either of these two proteins, we found Golgi mislocalization and extensive aberrant dendrite formation. Furthermore, overexpression of a mutated form of STRAD, underlying the disorder polyhydramnios, megalencephaly, and symptomatic epilepsy, characterized by abnormal brain development and intractable epilepsy, caused similar defects in Golgi localization and dendrite formation in adult-born neurons. Together, our findings reveal a role for Golgi repositioning in regulating the initial integration of adult-born DGCs.SIGNIFICANCE STATEMENT Since the discovery of the continuous generation of new neurons in the adult hippocampus, extensive effort was directed toward understanding the functional contribution of these newborn neurons to the existing hippocampal circuit and associated behaviors, while the molecular mechanisms controlling their early morphological integration are less well understood. Dentate granule cells (DGCs) have a single, complex, apical dendrite. The events leading adult-born DGCs' to transition from simple spindle-like morphology to mature dendrite morphology are largely unknown. We studied establishment of newborn DGCs dendritic pattern and found it was mediated by a signaling pathway regulating precise localization of the Golgi apparatus. Furthermore, this Golgi-associated mechanism for dendrite establishment might be impaired in a human genetic epilepsy syndrome, polyhydramnios, megalencephaly, and symptomatic epilepsy.
Collapse
|
21
|
Kumar P, Tan Y, Cahan P. Understanding development and stem cells using single cell-based analyses of gene expression. Development 2017; 144:17-32. [PMID: 28049689 DOI: 10.1242/dev.133058] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In recent years, genome-wide profiling approaches have begun to uncover the molecular programs that drive developmental processes. In particular, technical advances that enable genome-wide profiling of thousands of individual cells have provided the tantalizing prospect of cataloging cell type diversity and developmental dynamics in a quantitative and comprehensive manner. Here, we review how single-cell RNA sequencing has provided key insights into mammalian developmental and stem cell biology, emphasizing the analytical approaches that are specific to studying gene expression in single cells.
Collapse
Affiliation(s)
- Pavithra Kumar
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yuqi Tan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Hsieh J, Zhang CL. Neurogenesis in Cancun: where science meets the sea. Development 2017; 143:1649-54. [PMID: 27190035 DOI: 10.1242/dev.138412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 11/20/2022]
Abstract
In March 2016, meeting organizers Sebastian Jessberger and Hongjun Song brought together over 100 scientists from around the world to Cancun, Mexico to present the latest research on neurogenesis. The meeting covered diverse aspects of embryonic and adult neurogenesis with a focus on novel technologies, including chemogenetics and optogenetics, live cell two-photon imaging, cell fate reprogramming and human pluripotent stem cell models. This Meeting Review describes the exciting work that was presented and some of the emerging themes from the meeting.
Collapse
Affiliation(s)
- Jenny Hsieh
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| |
Collapse
|
23
|
Imaging Voltage in Genetically Defined Neuronal Subpopulations with a Cre Recombinase-Targeted Hybrid Voltage Sensor. J Neurosci 2017; 37:9305-9319. [PMID: 28842412 DOI: 10.1523/jneurosci.1363-17.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/09/2017] [Accepted: 08/16/2017] [Indexed: 12/16/2022] Open
Abstract
Genetically encoded voltage indicators create an opportunity to monitor electrical activity in defined sets of neurons as they participate in the complex patterns of coordinated electrical activity that underlie nervous system function. Taking full advantage of genetically encoded voltage indicators requires a generalized strategy for targeting the probe to genetically defined populations of cells. To this end, we have generated a mouse line with an optimized hybrid voltage sensor (hVOS) probe within a locus designed for efficient Cre recombinase-dependent expression. Crossing this mouse with Cre drivers generated double transgenics expressing hVOS probe in GABAergic, parvalbumin, and calretinin interneurons, as well as hilar mossy cells, new adult-born neurons, and recently active neurons. In each case, imaging in brain slices from male or female animals revealed electrically evoked optical signals from multiple individual neurons in single trials. These imaging experiments revealed action potentials, dynamic aspects of dendritic integration, and trial-to-trial fluctuations in response latency. The rapid time response of hVOS imaging revealed action potentials with high temporal fidelity, and enabled accurate measurements of spike half-widths characteristic of each cell type. Simultaneous recording of rapid voltage changes in multiple neurons with a common genetic signature offers a powerful approach to the study of neural circuit function and the investigation of how neural networks encode, process, and store information.SIGNIFICANCE STATEMENT Genetically encoded voltage indicators hold great promise in the study of neural circuitry, but realizing their full potential depends on targeting the sensor to distinct cell types. Here we present a new mouse line that expresses a hybrid optical voltage sensor under the control of Cre recombinase. Crossing this line with Cre drivers generated double-transgenic mice, which express this sensor in targeted cell types. In brain slices from these animals, single-trial hybrid optical voltage sensor recordings revealed voltage changes with submillisecond resolution in multiple neurons simultaneously. This imaging tool will allow for the study of the emergent properties of neural circuits and permit experimental tests of the roles of specific types of neurons in complex circuit activity.
Collapse
|
24
|
Methyl-CpG-Binding Protein MBD1 Regulates Neuronal Lineage Commitment through Maintaining Adult Neural Stem Cell Identity. J Neurosci 2017; 37:523-536. [PMID: 28100736 DOI: 10.1523/jneurosci.1075-16.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 10/31/2016] [Accepted: 11/22/2016] [Indexed: 01/09/2023] Open
Abstract
Methyl-CpG-binding domain 1 (MBD1) belongs to a family of methyl-CpG-binding proteins that are epigenetic "readers" linking DNA methylation to transcriptional regulation. MBD1 is expressed in neural stem cells residing in the dentate gyrus of the adult hippocampus (aNSCs) and MBD1 deficiency leads to reduced neuronal differentiation, impaired neurogenesis, learning deficits, and autism-like behaviors in mice; however, the precise function of MBD1 in aNSCs remains unexplored. Here, we show that MBD1 is important for maintaining the integrity and stemness of NSCs, which is critical for their ability to generate neurons. MBD1 deficiency leads to the accumulation of undifferentiated NSCs and impaired transition into the neuronal lineage. Transcriptome analysis of neural stem and progenitor cells isolated directly from the dentate gyrus of MBD1 mutant (KO) and WT mice showed that gene sets related to cell differentiation, particularly astrocyte lineage genes, were upregulated in KO cells. We further demonstrated that, in NSCs, MBD1 binds and represses directly specific genes associated with differentiation. Our results suggest that MBD1 maintains the multipotency of NSCs by restraining the onset of differentiation genes and that untimely expression of these genes in MBD1-deficient stem cells may interfere with normal cell lineage commitment and cause the accumulation of undifferentiated cells. Our data reveal a novel role for MBD1 in stem cell maintenance and provide insight into how epigenetic regulation contributes to adult neurogenesis and the potential impact of its dysregulation. SIGNIFICANCE STATEMENT Adult neural stem cells (aNSCs) in the hippocampus self-renew and generate neurons throughout life. We show that methyl-CpG-binding domain 1 (MBD1), a DNA methylation "reader," is important for maintaining the integrity of NSCs, which is critical for their neurogenic potency. Our data reveal a novel role for MBD1 in stem cell maintenance and provide insight into how epigenetic regulation preserves the multipotency of stem cells for subsequent differentiation.
Collapse
|
25
|
Zhang L, Tao W, Feng H, Chen Y. Transcriptional and Genomic Targets of Neural Stem Cells for Functional Recovery after Hemorrhagic Stroke. Stem Cells Int 2017; 2017:2412890. [PMID: 28133486 PMCID: PMC5241497 DOI: 10.1155/2017/2412890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/21/2016] [Indexed: 01/27/2023] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and it is widely believed that neural cell death occurs after exposure to blood metabolites or subsequently damaged cells. Neural stem cells (NSCs), which maintain neurogenesis and are found in subgranular zone and subventricular zone, are thought to be an endogenous neuroprotective mechanism for these brain injuries. However, due to the complexity of NSCs and their microenvironment, current strategies cannot satisfactorily enhance functional recovery after hemorrhagic stroke. It is well known that transcriptional and genomic pathways play important roles in ensuring the normal functions of NSCs, including proliferation, migration, differentiation, and neural reconnection. Recently, emerging evidence from the use of new technologies such as next-generation sequencing and transcriptome profiling has provided insight into our understanding of genomic function and regulation of NSCs. In the present article, we summarize and present the current data on the control of NSCs at both the transcriptional and genomic levels. Using bioinformatics methods, we sought to predict novel therapeutic targets of endogenous neurogenesis and exogenous NSC transplantation for functional recovery after hemorrhagic stroke, which could also advance our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Wenjing Tao
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
26
|
Johnson MB, Walsh CA. Cerebral cortical neuron diversity and development at single-cell resolution. Curr Opin Neurobiol 2016; 42:9-16. [PMID: 27888678 PMCID: PMC5316371 DOI: 10.1016/j.conb.2016.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/04/2016] [Indexed: 01/08/2023]
Abstract
Over a century of efforts to categorize the astonishing diversity of cortical neurons has relied on criteria of morphology, electrophysiology, ontology, and the expression of a few transcripts and proteins. The rapid development of single-cell RNA sequencing (scRNA-seq) adds genome-wide gene expression patterns to this list of criteria, and promises to reveal new insights into the transitions that establish neuronal identity during development, differentiation, activity, and disease. Comparing single neuron data to reference atlases constructed from hundreds of thousands of single-cell transcriptomes will be critical to understanding these transitions and the molecular mechanisms that drive them. We review early efforts, and discuss future challenges and opportunities, in applying scRNA-seq to the elucidation of neuronal subtypes and their development.
Collapse
Affiliation(s)
- Matthew B Johnson
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
27
|
Poirion OB, Zhu X, Ching T, Garmire L. Single-Cell Transcriptomics Bioinformatics and Computational Challenges. Front Genet 2016; 7:163. [PMID: 27708664 PMCID: PMC5030210 DOI: 10.3389/fgene.2016.00163] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
The emerging single-cell RNA-Seq (scRNA-Seq) technology holds the promise to revolutionize our understanding of diseases and associated biological processes at an unprecedented resolution. It opens the door to reveal intercellular heterogeneity and has been employed to a variety of applications, ranging from characterizing cancer cells subpopulations to elucidating tumor resistance mechanisms. Parallel to improving experimental protocols to deal with technological issues, deriving new analytical methods to interpret the complexity in scRNA-Seq data is just as challenging. Here, we review current state-of-the-art bioinformatics tools and methods for scRNA-Seq analysis, as well as addressing some critical analytical challenges that the field faces.
Collapse
Affiliation(s)
- Olivier B Poirion
- Epidemiology Program, University of Hawaii Cancer Center Honolulu, HI, USA
| | - Xun Zhu
- Epidemiology Program, University of Hawaii Cancer CenterHonolulu, HI, USA; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at ManoaHonolulu, HI, USA
| | - Travers Ching
- Epidemiology Program, University of Hawaii Cancer CenterHonolulu, HI, USA; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at ManoaHonolulu, HI, USA
| | - Lana Garmire
- Epidemiology Program, University of Hawaii Cancer Center Honolulu, HI, USA
| |
Collapse
|